Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Breast Cancer Res Treat ; 134(3): 1081-93, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22476851

RESUMEN

Tumor recurrence and metastasis result in an unfavorable prognosis for cancer patients. Recent studies have suggested that specific microRNAs (miRNAs) may play important roles in the development of cancer cells. However, prognostic markers and the outcome prediction of the miRNA signature in breast cancer patients have not been comprehensively assessed. The aim of this study was to identify miRNA biomarkers relating to clinicopathological features and outcome of breast cancer. A miRNA microarray analysis was performed on breast tumors of different lymph node metastasis status and with different progression signatures, indicated by overexpression of cyclin D1 and ß-catenin genes, to identify miRNAs showing a significant difference in expression. The functional interaction between the candidate miRNA, miR-30a, and the target gene, Vim, which codes for vimentin, a protein involved in epithelial-mesenchymal transition, was examined using the luciferase reporter assay, western blotting, and migration and invasion assays. The association between the decreased miR-30a levels and breast cancer progression was examined in a survival analysis. miR-30a negatively regulated vimentin expression by binding to the 3'-untranslated region of Vim. Overexpression of miR-30a suppressed the migration and invasiveness phenotypes of breast cancer cell lines. Moreover, reduced tumor expression of miR-30a in breast cancer patients was associated with an unfavorable outcome, including late tumor stage, lymph node metastasis, and worse progression (mortality and recurrence) (p < 0.05). In conclusion, these findings suggest a role for miR-30a in inhibiting breast tumor invasiveness and metastasis. The finding that miR-30a downmodulates vimentin expression might provide a therapeutic target for the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Vimentina/genética , Regiones no Traducidas 3' , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Biomarcadores de Tumor/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Humanos , MicroARNs/metabolismo , Persona de Mediana Edad , Metástasis de la Neoplasia/genética , Pronóstico , Interferencia de ARN , Adulto Joven
2.
J Cell Biochem ; 111(2): 274-83, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20506293

RESUMEN

Securin and γ-H2AX have been shown to regulate cell survival and genomic stability. However, it is still unknown how the expression and regulation of these proteins is altered following treatment with baicalein, a natural flavonoid extracted from the Scutellaria baicalensis root. In the present study, we investigate the possible roles of securin and γ-H2AX in baicalein-induced cancer cell death. Baicalein reduced cell viability in a variety of human cancer cell lines, including bladder, cervical, colon, and lung cancer cells. Interestingly, baicalein treatment (40-80 µM for 24 h) markedly inhibited securin expression, while the levels of γ-H2AX were elevated. Abnormal spindle formation and chromosomal segregation were induced by baicalein. Furthermore, wild type HCT116 cancer cells had a higher incidence of cytotoxicity and apoptosis than securin-null HCT116 cells following treatment with baicalein. In contrast, baicalein increased the levels of γ-H2AX to a similar extent in both cell types. Transfection with H2AX siRNA further increased baicalein-induced cell death. Additionally, blockade of the AKT pathway by treatment with wortmannin or AKT shRNA lowered the levels of γ-H2AX and enhanced cytotoxicity in baicalein-treated cells. Taken together, our findings suggest that the opposing effects of baicalein on securin and γ-H2AX levels may be involved in the regulation of cell viability and genomic stability by this compound.


Asunto(s)
Flavanonas/farmacología , Regulación de la Expresión Génica , Histonas/genética , Proteínas de Neoplasias/genética , Neoplasias/patología , Antioxidantes , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Flavanonas/uso terapéutico , Inestabilidad Genómica/efectos de los fármacos , Histonas/agonistas , Histonas/fisiología , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Securina
3.
Cancer Chemother Pharmacol ; 62(5): 799-808, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18193228

RESUMEN

BACKGROUND: The derivatives of 5,8-quinolinedione have been shown to exert anticancer activities. A new synthetic compound 7-chloro-6-piperidin-1-yl-quinoline-5,8-dione (designed as PT-262) derived from 6,7-dichloroquinoline-5,8-dione on its anticancer activity was investigated in this study. MATERIALS AND METHODS: PT-262 was synthesized as the following: triethylamine (0.56 ml, 5.1 mmol) was added dropwise to a solution of 6,7-dichloroquinoline-5,8-dione (1.00 g, 4.4 mmol) and piperidine (0.50 ml, 5.1 mmol) in 150 ml of benzene with stirring at room temperature for 5 min, and the solvent was removed using rotary evaporator to give a dark brown solid. PT-262 was purified by flash chromatography using 50% ethyl acetate/hexanes to elute that displayed as brown solids. To examine the induction of apoptosis following PT-262 treatment, the lung cancer cells were subjected to apoptotic cell observation, caspase activation, and mitochondrial functional assays. The protein levels of phosphorylated ERK and CDC2 after treatment with PT-262 were analyzed by Western blot. RESULTS: Treatment with 1-20 microM PT-262 for 24 h induced cytotoxicity via a concentration-dependent manner in human lung cancer cells. PT-262 induced the loss of mitochondrial membrane potential and elevated the caspase-3 activation and apoptosis. Interestingly, the phosphorylation of ERK was inhibited by PT-262. The IC50 value of ERK phosphorylation inhibition was approximate around 5 microM. Treatment with a specific MEK1/2 (the upstream of ERK) inhibitor, PD98059, increased the PT-262-induced cytotoxicity in lung cancer cells. Moreover, PT-262 did not alter the protein expression of tumor suppressor p53. PT-262 elicited the cytotoxicity and accumulated the G2/M fractions in both the p53-wild type and p53-null lung cancer cells. The mitosis-regulated protein levels of cyclin B1 and phospho-CDC2 at Thr14, Tyr15, and Thr161 were repressed by PT-262 in these cells. CONCLUSION: PT-262 suppresses the phosphorylation of ERK and CDC2 associated with proliferation inhibition via a p53-independent pathway in human lung cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Proteína Quinasa CDC2/antagonistas & inhibidores , Carcinoma/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Quinolonas/farmacología , Quinonas/farmacología , Proteína p53 Supresora de Tumor/fisiología , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Carcinoma/patología , Recuento de Células , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Potenciales de la Membrana/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Fosforilación , Quinolonas/química , Quinonas/química , Transducción de Señal/efectos de los fármacos
4.
Chem Biol Interact ; 176(1): 48-57, 2008 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-18760266

RESUMEN

Celecoxib, a clinical non-steroidal anti-inflammatory drug, displays anticarcinogenic and chemopreventive activities in human colorectal cancers, although the mechanisms of apoptosis by celecoxib are poorly understood. The existence of functional p53 but not securin in colorectal cancer cells was higher on the induction of cytotoxicity than the p53-mutational colorectal cancer cells following celecoxib treatment. The p53-wild type HCT116 cells were more susceptible to increase approximately 25% cell death than the p53-null HCT116 cells after treatment with 100 microM celecoxib for 24 h. Transfection with a small interfering RNA of p53 reduced the celecoxib-induced cytotoxicity in the RKO (p53-wild type) colorectal cancer cells. Celecoxib (80-100 microM for 24 h) significantly increased total p53 proteins and the phosphorylated p53 proteins at serine-15, -20, -46, and -392 in RKO cells. However, the phospho-p53 (serine-15, -20, and -392) proteins were presented on the nuclei of cells but the phospho-p53 (serine-46) protein was located on the cytoplasma of apoptotic cells following treatment with celecoxib. Interestingly, the p53 up-regulated modulator of apoptosis (PUMA) protein, which located on the mitochondria, was induced by celecoxib in the p53-functional colorectal cancer cells but not in the p53-mutational cells. Together, this study provides the first time that celecoxib induces the various phosphorylated sites of p53 and activates p53-PUMA pathway, which potentiates the apoptosis induction in human colorectal cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas Proto-Oncogénicas/metabolismo , Pirazoles/farmacología , Sulfonamidas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Celecoxib , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación/genética , Proteínas de Neoplasias/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Transporte de Proteínas/efectos de los fármacos , Securina
5.
Mol Cancer Ther ; 6(11): 3039-48, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18025287

RESUMEN

The bioactive flavonoid baicalein has been shown to have in vitro growth-inhibitory activity in human cancer cells, although the mechanism of action is poorly understood. Baicalein (40-80 mumol/L for 24 h) more effectively induced cytotoxicity compared with other flavonoids (baicalin, catechin, genistein, quercetin, and rutin) in bladder cancer cells. Baicalein induced cell proliferation inhibition and apoptosis. The levels of cyclin B1 and phospho-CDC2 (Thr(161)) were reduced, whereas the G(2)-M phases were elevated by baicalein. Treatment of CDC2 kinase or CDC25 phosphatase inhibitors augments the baicalein-induced cytotoxicity. A variety of human bladder cancer cell lines expressed survivin proteins, which were located on the mitotic phases and regulated mitotic progression. Baicalein markedly reduced survivin protein expression. Transfection of a survivin small interfering RNA diminished the level of survivin proteins and increased the baicalein-mediated cell death. Overexpression of survivin enhanced cell proliferation and resisted the baicalein-induced cytotoxicity. Interestingly, baicalein induced the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and AKT. SB203580, a specific p38 MAPK inhibitor, attenuated proliferation inhibition and restored the protein levels of phospho-CDC2 (Thr(161)) and survivin in the baicalein-exposed cells; conversely, blockade of AKT activation enhanced cytotoxicity and the reduction of phospho-CDC2 (Thr(161)) and survivin proteins. As a whole, these findings provide that the opposite role of p38 MAPK and AKT regulates CDC2 kinase and survivin and the inhibition of CDC2-survivin pathway by baicalein contributes to apoptosis and proliferation retardation in cancer cells.


Asunto(s)
Proteína Quinasa CDC2/antagonistas & inhibidores , Flavanonas/farmacología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias de la Vejiga Urinaria/enzimología , Neoplasias de la Vejiga Urinaria/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina B/metabolismo , Ciclina B1 , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Flavanonas/química , Flavonoides/farmacología , Fase G2/efectos de los fármacos , Humanos , Proteínas Inhibidoras de la Apoptosis , Mitosis/efectos de los fármacos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Survivin
6.
Autophagy ; 13(1): 187-200, 2017 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-27846374

RESUMEN

Selective macroautophagy/autophagy plays a pivotal role in the processing of foreign pathogens and cellular components to maintain homeostasis in human cells. To date, numerous studies have demonstrated the uptake of nanoparticles by cells, but their intracellular processing through selective autophagy remains unclear. Here we show that carbon-based nanodiamonds (NDs) coated with ubiquitin (Ub) bind to autophagy receptors (SQSTM1 [sequestosome 1], OPTN [optineurin], and CALCOCO2/NDP52 [calcium binding and coiled-coil domain 2]) and are then linked to MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3) for entry into the selective autophagy pathway. NDs are ultimately delivered to lysosomes. Ectopically expressed SQSTM1-green fluorescence protein (GFP) could bind to the Ub-coated NDs. By contrast, the Ub-associated domain mutant of SQSTM1 (ΔUBA)-GFP did not bind to the Ub-coated NDs. Chloroquine, an autophagy inhibitor, prevented the ND-containing autophagosomes from fusing with lysosomes. Furthermore, autophagy receptors OPTN and CALCOCO2/NDP52, involved in the processing of bacteria, were found to be involved in the selective autophagy of NDs. However, ND particles located in the lysosomes of cells did not induce mitotic blockage, senescence, or cell death. Single ND clusters in the lysosomes of cells were observed in the xenografted human lung tumors of nude mice. This study demonstrated for the first time that Ub-coated nanoparticles bind to autophagy receptors for entry into the selective autophagy pathway, facilitating their delivery to lysosomes.


Asunto(s)
Autofagia , Nanodiamantes/química , Ubiquitina/química , Células A549 , Animales , Muerte Celular , Línea Celular Tumoral , Senescencia Celular , Proteínas Fluorescentes Verdes/química , Humanos , Neoplasias Pulmonares/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Desnudos , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/química , Trasplante de Neoplasias , Proteínas del Tejido Nervioso/química , Proteínas Nucleares/química , Unión Proteica , Receptores Citoplasmáticos y Nucleares/química , Proteínas Recombinantes/química , Proteína Sequestosoma-1/química
7.
Chem Biol Interact ; 203(2): 412-22, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23523951

RESUMEN

Gefitinib, a tyrosine kinase inhibitor of the epidermal growth factor receptor (EGFR), has been used to treat numerous cancers; however, evidence has shown that cancer cells can become resistant to gefitinib during therapy. Here, we report a human proto-oncogene, securin, which displays resistance to death in cancer cells. Gefitinib treatment decreases securin levels at the protein level by inducing protein instability but did not affect on the securin gene expression. Treatment with gefitinib induced cytotoxicity in various human cancer cell types, including RKO (colon cancer), A549 (lung cancer), BFTC905 (bladder cancer), MCF7 (breast cancer) and A375 (skin cancer). BFTC905 and A549 cells expressed relatively high levels of the phosphorylated and total EGFR proteins; however, A375, MCF7 and RKO cells did not markedly express these proteins. Moreover, following treatment with gefitinib, the securin-wild type cancer cells were more resistant to apoptotic induction than the securin-null cancer cells. Surprisingly, both the securin-wild type and securin-null cancer cells expressed the EGFR protein at similar levels. Treatment with gefitinib induced mitochondrial dysfunction, cytochrome c release, caspase-3 activation and poly (ADP-ribose) polymerase protein cleavage, indicating that apoptosis occurred in these cancer cells. The transfection of a GPF-securin expression vector increased both the proliferation rates and resistance to gefitinib-induced death in these cancer cells. Taken together, these findings demonstrate that the presence of securin promotes resistance to gefitinib-induced apoptosis via an EGFR-independent pathway in human cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos , Proteínas de Neoplasias/metabolismo , Quinazolinas/farmacología , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Receptores ErbB/metabolismo , Gefitinib , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de Neoplasias/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proto-Oncogenes Mas , Securina , Transducción de Señal/efectos de los fármacos
8.
Biochem Pharmacol ; 81(7): 856-65, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21276421

RESUMEN

The 5,8-quinolinediones are precursors for producing multiple types of bioactive products. In this study, we investigated a new compound derived from 5,8-quinolinediones, 7-chloro-6-piperidin-1-yl-quinoline-5,8-dione (designated as PT-262), which markedly induced cytoskeleton remodeling and migration inhibition in lung carcinoma cells. Comparison with various cytoskeleton inhibitors, including paclitaxel, colchicine and phallacidin, the cell morphology following treatment with PT-262 was similar to phallacidin on the cell elongation and abnormal actin polymerization. However, PT-262 did not directly bind to actin filaments. ROCK (Rho-associated coiled-coil forming protein kinase) is a downstream effector of RhoA to mediate the phosphorylation of myosin light chain (MLC) and cytoskeleton reorganization. The RhoA-ROCK-MLC pathway has been shown to promote cancer cell migration and metastasis. Interestingly, PT-262 was more effective on inhibiting ROCK kinase activities than specific ROCK inhibitors Y-27632 and H-1152. PT-262 induced cytoskeleton remodeling and migration inhibition in A549 lung carcinoma cells. The total MLC and phosphorylated MLC proteins and stress fibers were blocked after treatment with PT-262. Nonetheless, the RhoA protein and GTPase activity were not altered by PT-262. A computational model suggests that PT-262 interacts with the ATP-binding site of ROCK protein. Together, these findings demonstrate that PT-262 is a new ROCK inhibitor.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Quinolonas/farmacología , Quinonas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Citoesqueleto/fisiología , Humanos , Microscopía de Fuerza Atómica , ARN Interferente Pequeño
9.
Chem Biol Interact ; 188(3): 535-45, 2010 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-20708607

RESUMEN

Oxaliplatin, a platinum derivative cancer drug, has been used for treating human colorectal cancers. Survivin has been proposed as a cancer target, which highly expressed in most cancer cells but not normal adult cells. In this study, we investigated the regulation of survivin expression by exposure to oxaliplatin in human colon cancer cells. Oxaliplatin (3-9µM for 24h) markedly induced cytotoxicity, proliferation inhibition and apoptosis in the human RKO colon cancer cells. The survivin protein expression of RKO cells is dramatically reduced by oxaliplatin; however, the survivin gene expression is slightly altered. The survivin blockage of oxaliplatin elevated caspase-3 activation and apoptosis in RKO cells. Over-expression of survivin proteins by transfection with a survivin-expressed vector resisted the oxaliplatin-induced cancer cell death. Meantime, oxaliplatin elicited the phosphorylation of p38 mitogen-activated protein (MAP) kinase. SB202190, a specific p38 MAP kinase inhibitor, restored the survivin protein level and attenuated oxaliplatin-induced cancer cell death. In addition, oxaliplatin increased the levels of phospho-p53 (Ser-15) and total p53 proteins. Inhibition of p53 expression by a specific p53 inhibitor pifithrin-α reduced the phosphorylated p38 MAP kinase and active caspase-3 proteins in the oxaliplatin-exposed RKO cells. In contrast, SB202190 did not alter the oxaliplatin-induced p53 protein level. Furthermore, treatment with a specific proteasome inhibitor MG132 restored survivin protein level in the oxaliplatin-treated colon cancer cells. Taken together, our results demonstrate for the first time that survivin is down-regulated by p38 MAP kinase and proteasome degradation pathway after treatment with oxaliplatin in the human colon cancer cells.


Asunto(s)
Neoplasias del Colon/patología , Regulación hacia Abajo/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Compuestos Organoplatinos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Proteínas Inhibidoras de la Apoptosis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Oxaliplatino , Fosforilación/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Piridinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Survivin
10.
Toxicol Appl Pharmacol ; 222(1): 97-104, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17540426

RESUMEN

Cancer cells express survivin that facilitates tumorigenesis. Celecoxib has been shown to reduce human colorectal cancers. However, the role and regulation of survivin by celecoxib in colorectal carcinoma cells remain unclear. Treatment with 40-80 muM celecoxib for 24 h induced cytotoxicity and proliferation inhibition via a concentration-dependent manner in RKO colorectal carcinoma cells. Celecoxib blocked the survivin protein expression and increased the phosphorylation of H2AX at serine-193 (gamma-H2AX). The survivin gene knockdown by transfection with a survivin siRNA revealed that the loss of survivin correlated with the expression of gamma-H2AX. Meanwhile, celecoxib increased caspase-3 activation and apoptosis. Celecoxib activated the phosphorylation of p38 mitogen-activated protein (MAP) kinase. The phosphorylated proteins of p38 MAP kinase and gamma-H2AX were observed in the apoptotic cells. SB203580, a specific p38 MAP kinase inhibitor, protected the survivin protein expression and decreased the levels of gamma-H2AX and apoptosis in the celecoxib-exposed cells. The blockade of survivin expression increased the celecoxib-induced cytotoxicity; conversely, overexpression of survivin by transfection with a survivin-expressing vector raised the cancer cell proliferation and resisted the celecoxib-induced cell death. Our results provide for the first time that p38 MAP kinase participates in the down-regulation of survivin and subsequently induces the activation of gamma-H2AX for mediating apoptosis following treatment with celecoxib in human colorectal cancer cells.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Histonas/biosíntesis , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas de Neoplasias/biosíntesis , Pirazoles/farmacología , Sulfonamidas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Celecoxib , Recuento de Células , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Fosforilación , ARN Interferente Pequeño/farmacología , Survivin , Transfección
11.
Mol Pharmacol ; 69(1): 154-64, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16219911

RESUMEN

Survivin and securin proteins are overexpressed in most cancer cells that have been shown to regulate mitotic progression. In this study, we investigated the roles of survivin and securin on cytochalasin B, a cytokinesis blocker mediating the cytotoxicity and cell growth inhibition in human cancer cells. The human lung carcinoma cell lines A549 and H1299 highly expressed survivin proteins in mitosis and concentrated on the midbodies during cytokinesis. Cytochalasin B significantly decreased cell survival, inhibited cell growth, increased the levels of G(2)/M fractions, and induced binuclei formation in lung carcinoma cells; however, the survivin proteins were concentration-dependently increased by 1 to 5 mug/ml cytochalasin B for 24 h. It is noteworthy that the expression of securin proteins was decreased in cytochalasin B-treated lung carcinoma cells. Transfection of 20 to 40 nM survivin siRNA for 48 h significantly induced the formation of multiple nuclei and apoptosis but decreased the levels of survivin and securin proteins in A549 cells. Cotreatment with survivin small interfering RNA (siRNA) and cytochalasin B increased the cytotoxicity and cell growth inhibition. In addition, the securin-null colorectal carcinoma cells were more susceptible to the cytotoxicity after cytochalasin B and survivin siRNA treatments than the securin-wild-type cells. As a whole, our results indicate that the inhibition of survivin and securin protein expression may increase the cell death and growth inhibition after cytochalasin B treatment in human cancer cells.


Asunto(s)
Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Citocalasina B/farmacología , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Ciclo Celular , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas Inhibidoras de la Apoptosis , Neoplasias Pulmonares/patología , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , ARN Interferente Pequeño , Securina , Survivin
12.
J Biol Chem ; 279(53): 55875-85, 2004 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-15456784

RESUMEN

Quercetin, a ubiquitous bioactive plant flavonoid, has been shown to inhibit the proliferation of cancer cells. However, the regulation of survivin and p53 on the quercetin-induced cell growth inhibition and apoptosis in cancer cells remains unclear. In this study, we investigated the roles of survivin and p53 in the quercetin-treated human lung carcinoma cells. Quercetin (20-80 mum for 24 h) induced the cytotoxicity and apoptosis in both A549 and H1299 lung carcinoma cells in a concentration-dependent manner. Additionally, quercetin inhibited the cell growth, increased the fractions of G(2)/M phase, and raised the levels of cyclin B1 and phospho-cdc2 (threonine 161) proteins. Moreover, quercetin induced abnormal chromosome segregation in H1299 cells. The survivin proteins were highly expressed in mitotic phase and were located on the midbody of cytokinesis; however, the survivin proteins were increased and concentrated on the nuclei following quercetin treatment in the lung carcinoma cells. Transfection of a survivin antisense oligodeoxynucleotide enhanced the quercetin-induced cell growth inhibition and cytotoxicity. Subsequently, quercetin increased the levels of total p53 (DO-1), phospho-p53 (serine 15), and p21 proteins, which were translocated to the nuclei in A549 cells. Treatment with a specific p53 inhibitor, pifithrin-alpha, or transfection of a p53 antisense oligodeoxynucleotide enhanced the cytotoxicity of the quercetin-treated cells. Furthermore, transfection of a small interfering RNA of p21 enhanced the quercetin-induced cell death in A549 cells. Together, our results suggest that survivin can reduce the cell growth inhibition and apoptosis, and p53 elevates the p21 level, which may attenuate the cell death in the quercetin-treated human lung carcinoma cells.


Asunto(s)
Apoptosis , Carcinoma/patología , Proteínas de Ciclo Celular/metabolismo , Neoplasias Pulmonares/patología , Proteínas Asociadas a Microtúbulos/fisiología , Quercetina/farmacología , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/metabolismo , Benzotiazoles , Western Blotting , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Ciclina B/metabolismo , Ciclina B1 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , ADN/química , Técnica del Anticuerpo Fluorescente Indirecta , Fase G2 , Humanos , Proteínas Inhibidoras de la Apoptosis , Masculino , Microscopía de Contraste de Fase , Persona de Mediana Edad , Mitosis , Modelos Biológicos , Proteínas de Neoplasias , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/farmacología , Fosforilación , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo , Survivin , Tiazoles/metabolismo , Treonina/metabolismo , Factores de Tiempo , Tolueno/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA